EP4149947A1 - Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein - Google Patents

Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein

Info

Publication number
EP4149947A1
EP4149947A1 EP21730723.0A EP21730723A EP4149947A1 EP 4149947 A1 EP4149947 A1 EP 4149947A1 EP 21730723 A EP21730723 A EP 21730723A EP 4149947 A1 EP4149947 A1 EP 4149947A1
Authority
EP
European Patent Office
Prior art keywords
crystalline form
degrees
theta
peaks
diffraction pattern
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21730723.0A
Other languages
German (de)
English (en)
French (fr)
Inventor
Jincong Zhuo
Ganfeng Cao
Andrew Paul Combs
Qun Li
Huaping Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Prelude Therapeutics Inc
Original Assignee
Prelude Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Prelude Therapeutics Inc filed Critical Prelude Therapeutics Inc
Publication of EP4149947A1 publication Critical patent/EP4149947A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D515/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D515/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D515/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D515/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D515/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D515/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • Apoptosis (programmed cell death) is a highly conserved cellular process that is required for embryonic development and normal tissue homeostasis (Ashkenazi A. et al, Nat. Rev. Drug Discov. 2017, 16, 273-284).
  • Apoptotic-type cell death involves morphological changes such as condensation of the nucleus, DNA fragmentation as well as biochemical phenomena such as the activation of caspases which cause damage to key structural components of the cell, resulting in its disassembly and death.
  • Regulation of the process of apoptosis is complex and involves the activation or repression of several intracellular signaling pathways (Cory S. et al, Nature Review Cancer 2002, 2, 647-656; Thomas L. W. etal, FEBS Lett. 2010, 584, 2981-2989; Adams J. M. et al., Oncogene 2007, 26, 1324-1337)
  • the Bcl-2 protein family which includes both pro-apoptotic and anti-apoptotic members, plays a pivotal role in the regulation of the apoptosis process (Youle R. J. etal, Nat. Rev. Mol. Cell Biol. 2008, 9, 47-59; Kelly G. L. etal, Adv. Cancer Res. 2011, 111, 39-96).
  • Bcl-2, Bel -XL, Bcl-W, Mcl-1 and Al are anti-apoptotic proteins and they share a common BH regions.
  • the pro-apoptotic family members are divided into two groups.
  • the multiregion pro-apoptotic proteins such as Bax, Bak and Bok
  • the BH3-only proteins are proposed to share homology in the BH3 region only.
  • Members of BH3-only proteins include Bad, Bim, Bid, Noxa, Puma, Bik/Blk, Bmf, Hrk/DP5, Beclin-1 and Mule (Xu G. et al, Bioorg. Med. Chem. 2017, 25, 5548-5556; Hardwick J. M. etal, Cell. 2009, 138, 404; Reed J. C., Ceil Death Differ. 2018, 2.5, 3-6; Kang M. H. etal, Clin Cancer Res 2009, 15, 1126-1132).
  • the pro-apoptotic members upon activation, form a homo-oligomer in the outer mitochondrial membrane that leads to pore formation and the escape of mitochondrial contents, a step into triggering apoptosis.
  • Antiapoptotic members of the Bcl-2 family (such as Bcl-2, Bel -XL, and Mcl-1) block the activity of BAX and BAK. In normal cells, this process is tightly regulated. Abnormal cells can dysregulate this process to avoid cell death.
  • One of the ways that cancer cells can accomplish this is by upregulating the antiapoptotic members of the Bcl-2 family of proteins.
  • Aberrant expression or function of the proteins responsible for apoptotic signaling contributes to numerous human pathologies including auto-immune diseases, neurodegeneration (such as Parkinson’s disease, Alzheimer’s disease and ischaemia), inflammatory diseases, viral infections and cancer (such as colon cancer, breast cancer, small-cell lung cancer, non-small-cell lung cancer, bladder cancer, ovarian cancer, prostate cancer, chronic lymphoid leukemia, lymphoma, myeloma, acute myeloid leukemia, pancreatic cancer, etc.) (Hanahan D. et cil, Cell 2000, 100. 57-70).
  • it is prospective to target key apoptosis regulators for cancer treatment (Kale J. et al, Cell Death Differ. 2018, 25, 65-80: Vogler M. et al, Cell Death Differ. 2009, 16, 360-367).
  • Mcl-1 Myeloid Cell Leukemia-1
  • Mcl-1 has the distinct trait of being essential for embryonic development as well as the survival of all hematopoietic lineages and progenitor populations. Mcl-1 is one of the most common genetic aberrations in human cancer and is highly expressed in many tumor types. Mcl-1 overexpression in human cancers is associated with high tumor grade and poor survival (Beroukhim R. et al, Nature 2010, 463, 899-905).
  • Mcl-1 overexpression prevents cancer cells from undergoing programmed cell death (apoptosis), allowing the cells to survive despite widespread genetic damage. Further, its amplification is associated with both intrinsic and acquired resistance to a wide variety of antitumorigenic agents including chemotherapeutic agents such as microtubule binding agents, paclitaxel and gemcitabine, as well as apoptosis-inducing agents such as TRAIL, the Bcl-2 inhibitor, venetoclax, and the Bcl-2/Bcl-XL dual inhibitor navitoclax.
  • chemotherapeutic agents such as microtubule binding agents, paclitaxel and gemcitabine
  • apoptosis-inducing agents such as TRAIL, the Bcl-2 inhibitor, venetoclax, and the Bcl-2/Bcl-XL dual inhibitor navitoclax.
  • the disclosure is also directed to crystalline forms of [(3R,6R,7S,8E,22S)-6'-Chloro- 12, 12-dimethyl- 13,15,15-trioxo-spiro[ 11 ,20-dioxa- 15 -thia- 1,14- diazatetracyclo[14.7.2.03,6.019,24]-pentacosa-8,16,18,24-tetraene-22,1'-tetralin]-7-yl] N,N- dimethylcarbamate, i. e.. the compound of Formula I,
  • compositions containing such forms are also described.
  • the disclosure is also directed to choline, benzathine, imidazole, piperazine, piperidine, (S)-(-)- ⁇ -methylbenzylamine, ethylenediamine, potassium, and 4-((2-aminoethyl)amino)-4- methylpentan-2-one salts of Formula I.
  • Figure 1 shows an XRPD of Formula I-Form I.
  • Figure 2 shows a DSC thermogram of Formula I-Form I.
  • Figure 3 shows a TGA profile of Formula I-Form I.
  • Figures 4A and 4B show a DVS profile of Formula I-Form I.
  • Figure 5 shows an XRPD of Formula I-Form I before (top) and after (bottom) DVS.
  • Figure 6 shows an XRPD of Formula I-Form II.
  • Figure 7 shows a DSC thermogram of Formula I-Form II.
  • Figure 8 shows an XRPD of a choline salt of Formula I.
  • Figure 9 shows a DSC thermogram of a choline salt of Formula I.
  • Figure 10 shows a TGA profile of a choline salt of Formula I.
  • Figure 11 shows an NMR spectrum (600 MHz in CDCl 3 ) of a choline salt of Formula I.
  • Figure 12 shows an XRPD of a benzathine salt of Formula I.
  • Figure 13 shows a DSC thermogram of a benzathine salt of Formula I.
  • Figure 14 shows a TGA profile of a benzathine salt of Formula I.
  • Figure 15 shows an NMR spectrum (600 MHz in CDCl 3 ) of a benzathine salt of Formula I.
  • Figure 16 shows an XRPD of an imidazole salt of Formula I.
  • Figure 17 shows a DSC thermogram of an imidazole salt of Formula I.
  • Figure 18 shows a TGA profile of an imidazole salt of Formula I.
  • Figure 19 shows an NMR spectrum (600 MHz in CDCl 3 ) of an imidazole salt of Formula I.
  • Figure 20 shows an XRPD of a piperazine salt of Formula I (Form 1).
  • Figure 20A shows an XRPD of a piperazine salt of Formula I (Form 2)
  • Figure 20B shows an XRPD of a piperazine salt of Formula I (Form 3)
  • Figure 21 shows a DSC thermogram of a piperazine salt of Formula I (Form 1).
  • Figure 21A shows a DSC thermogram of a piperazine salt of Formula I (Form 2).
  • Figure 22 shows a TGA profile of a piperazine salt of Formula I (Form 1).
  • Figure 23 shows an NMR spectrum (600 MHz in CDCl 3 ) of a piperazine salt of Formula I (Form 1).
  • Figure 24 shows an XRPD of a piperidine salt of Formula I (Form 1).
  • Figure 24A shows an XRPD of a piperidine salt of Formula I (Form 2).
  • Figure 25 shows a DSC thermogram of a piperidine salt of Formula I (Form 1).
  • Figure 26 shows a TGA profile of a piperidine salt of Formula I (Form 1).
  • Figure 27 shows an NMR spectrum (600 MHz in CDCl 3 ) of a piperidine salt of Formula I (Form 1).
  • Figure 28 shows an XRPD of a potassium salt of Formula I.
  • Figure 29 shows a DSC thermogram of a potassium salt of Formula I.
  • Figure 30 shows an XRPD of a (S -(-)- ⁇ -Methylbenzylamine salt of Formula I.
  • Figure 31 shows a DSC thermogram of a (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I.
  • Figure 32 shows an XRPD of an ethylenediamine salt of Formula I (Form 1).
  • Figure 32A shows an XRPD of an ethylenediamine salt of Formula I (Form 2).
  • Figure 33 shows NMR Spectrum of an ethylenediamine salt of Formula I (Form 1).
  • Figure 34 shows an XRPD of a 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I.
  • Figure 35 shows a DSC thermogram of a 4-((2-aminoethyl)amino)-4-methylpentan-2- one salt of Formula I.
  • Figure 36 shows a TGA profile of a 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I.
  • Figure 37 shows an NMR spectrum (600 MHz in CDCl 3 ) of a 4-((2-aminoethyl)amino)- 4-methylpentan-2-one salt of Formula I.
  • compositions and methods which are described herein in the context of separate aspects, may also be provided in combination in a single aspect.
  • “Pharmaceutically acceptable” means approved or approvable by a regulatory agency of the Federal or a state government or the corresponding agency in countries other than the United States, or that is listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, e.g., in humans.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the disclosure that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • such salts are non-toxic may be inorganic or organic acid addition salts and base addition salts.
  • such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid,
  • 3-(4-hydroxybenzoyl)benzoic acid cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxy ethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylic acid, glucoheptonic acid,
  • 3-phenylpropionic acid trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N- methylglucamine and the like.
  • a metal ion e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion
  • coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N- methylglucamine and the like.
  • Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
  • non-toxic organic or inorganic acids such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
  • a “pharmaceutically acceptable excipient” refers to a substance that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to a subject, such as an inert substance, added to a pharmacological composition or otherwise used as a vehicle, carrier, or diluent to facilitate administration of an agent and that is compatible therewith.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
  • a “solvate” refers to a physical association of a compound of Formula I with one or more solvent molecules.
  • Subject includes humans.
  • the terms “human,” “patient,” and “subject” are used interchangeably herein.
  • Treating” or “treatment” of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment “treating” or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, “treating” or “treatment” refers to delaying the onset of the disease or disorder.
  • isotopic variant refers to a compound that contains proportions of isotopes at one or more of the atoms that constitute such compound that is greater than natural abundance.
  • an “isotopic variant” of a compound can be radiolabeled, that is, contain one or more radioactive isotopes, or can be labeled with non-radioactive isotopes such as for example, deuterium ( 2 H or D), carbon- 13 ( 13 C), nitrogen- 15 ( 15 N), or the like.
  • any hydrogen may be 2 H/D
  • any carbon may be 13 C
  • any nitrogen may be 15 N, and that the presence and placement of such atoms may be determined within the skill of the art.
  • isomers compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed “isomers.” Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers,” for example, diastereomers, enantiomers, and atropisomers.
  • the compounds of this disclosure may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)-or (S)-stereoisomers at each asymmetric center, or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include all stereoisomers and mixtures, racemic or otherwise, thereof.
  • the disclosure is directed to a crystalline form of the compound of Formula I,
  • the disclosure is directed to crystalline form I of the compound of Formula I (Formula I-Form I).
  • Formula I-Form I is substantially free of any other solid form of Formula I.
  • Formula I-Form I exhibits an XRPD substantially as shown in Figure 1.
  • the XRPD of Formula I-Form I shown in Figure 1 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 1: Table 1.
  • Formula I-Form I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 1. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 1 above.
  • Formula I-Form I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 1 above.
  • Formula I-Form I is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 1 above. In other aspects, Formula I-Form I is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 1 above.
  • Formula I-Form I is characterized by an XRPD pattern comprising a peak 11.2, 13.9, 17.1, 17.7, and 20.8 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, Formula I-Form I is characterized by an XRPD pattern comprising peaks at 9.4, 11.2, 13.9, 17.1, and 17.7 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, Formula I-Form I is characterized by an XRPD pattern comprising peaks at 17.1, 17.7, 20.8, and 21.9 degrees ⁇ 0.2 degree 2-theta.
  • Formula I-Form I is characterized by an XRPD pattern comprising peaks at 13.9, 17.1, 17.7, 20.8, and 21.9 degrees ⁇ 0.2 degree 2-theta. In other embodiments, Formula I-Form I is characterized by an XRPD pattern comprising peaks at 11.2, 13.9, 17.1, 17.7, 20.8, 21.9, and 25.0 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, Formula I-Form I is characterized by an XRPD pattern comprising peaks at 9.4, 11.2, 13.9, 17.1, 17.7, 20.8, 21.9, 25.0, and 27.8 degrees ⁇ 0.2 degree 2-theta.
  • Formula I-Form I is characterized by an XRPD pattern comprising peaks at two or more of 9.4, 11.2, 13.9, 17.1, 17.7, 20.8, 21.9, 25.0, and 27.8 degrees ⁇ 0.2 degrees 2-theta.
  • Formula I-Form I can be characterized by a DSC thermogram substantially as shown in Figure 2. As Figure 2 shows, Formula I-Form I produced an endothermic peak at 81.29 °C, with a peak onset temperature of 66.26 °C, and an enthalpy of melting of 36.11 J/g, when heated at a rate of 10°C/min. In some embodiments of the present disclosure, Formula I-Form I is characterized by a DSC thermogram comprising an endothermic peak at about 81°C. In other embodiments of the present disclosure, Formula I-Form I is characterized by a DSC enthalpy of melting of about 36 J/g.
  • Formula I-Form I can be characterized by a TGA profile substantially as shown in Figure 3 when heated at a rate of 20°C/min. As Figure 3 shows, Formula I-Form I lost about 76% of its weight upon heating to about 430°C.
  • Formula I-Form I is characterized by an XRPD pattern comprising peaks at one or more of 9.4, 11.2, 13.9, 17.1, 17.7, 20.8, 21.9, 25.0, and 27.8 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 81°C when heated at a rate of 10°C/min.
  • the disclosure is directed to crystalline Form II of the compound of Formula I (Formula I-Form II).
  • Formula I-Form II is substantially free of any other solid form of Formula I.
  • Formula I-Form II exhibits an XRPD substantially as shown in Figure 6.
  • the XRPD of Formula I-Form II shown in Figure 6 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 2:
  • Formula I-Form II is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 2. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 2 above.
  • Formula I-Form II is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 2 above. In other aspects,
  • Formula I-Form II is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 2 above. In other aspects, Formula I-Form II is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 2 above.
  • Formula I-Form II is characterized by an XRPD pattern comprising a peak 9.2, 21.7, and 30.5 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, Formula I-Form II is characterized by an XRPD pattern comprising peaks at 9.2, 12.6, 17.4, and 30.5 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, Formula I-Form II is characterized by an XRPD pattern comprising peaks at 17.4, 18.1, 19.3, 19.8, and 21.7 degrees ⁇ 0.2 degree 2- theta. In other embodiments, Formula I-Form II is characterized by an XRPD pattern comprising peaks at 17.4, 18.1, 19.3, 19.8, and 30.5degrees ⁇ 0.2 degree 2-theta.
  • Formula I-Form II is characterized by an XRPD pattern comprising peaks at 12.6, 17.4, 18.1, 19.3, 19.8, 21.7, 28.6, and 30.5degrees ⁇ 0.2 degree 2-theta. . In other embodiments, Formula I-Form II is characterized by an XRPD pattern comprising peaks at 9.2, 12.6, 17.4,
  • Formula I-Form II is characterized by an XRPD pattern comprising peaks at two or more of 9.2, 12.6, 17.4, 18.1, 19.3, 19.8, 21.7, 28.6,
  • Formula I-Form II can be characterized by a DSC thermogram substantially as shown in Figure 7. As Figure 7 shows, Formula I-Form II produced an endothermic peak at 68.06 °C, with a peak onset temperature of 64.20 °C, and an enthalpy of melting of 22.71 J/g, followed by produced an endothermic peak at 91.90 °C, with a peak onset temperature of 85.85 °C, and an enthalpy of melting of 114.7 J/g, when heated at a rate of 10°C/min. In some embodiments of the present disclosure, Formula I-Form II is characterized by a DSC thermogram comprising an endothermic peak at about 68°C.
  • Formula I-Form II is characterized by a DSC enthalpy of melting of about 23 J/g. In other embodiments, Formula I-Form II is characterized by a DSC thermogram comprising an endothermic peak at about 92°C. In other embodiments of the present disclosure, Formula I-Form II is characterized by a DSC enthalpy of melting of about 115 J/g.
  • Formula I-Form II is characterized by an XRPD pattern comprising peaks at one or more of 9.2, 12.6, 17.4, 18.1, 19.3, 19.8, 21.7, 28.6,
  • the disclosure is directed to a choline salt of a compound of Formula I, having the formula IA:
  • the disclosure is directed to a crystalline form of the choline salt of the compound of Formula I.
  • the choline salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the choline salt of formula I exhibits an XRPD substantially as shown in Figure 8.
  • the XRPD of the choline salt of formula I shown in Figure 8 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 3:
  • the choline salt of Formula I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 3. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 3 above.
  • the choline salt of Formula I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 3 above.
  • the choline salt of Formula I is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 3 above. In other aspects, the choline salt of Formula I is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 3 above.
  • the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at 19.4, and 20.0 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at 18.5,
  • the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at 18.5, 19.4, 20.0, 22.6, and 24.7 degrees ⁇ 0.2 degree 2-theta. In other embodiments, the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at 13.3, 18.5, 19.4, 20.0, and 22.6 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at 13.3, 18.5, 19.4, 20.0, 22.6, and 24.7 degrees ⁇ 0.2 degree 2- theta. . In other embodiments, the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at 9.9, 13.3, 18.5, 19.4, 20.0, 22.6, and 24.7 degrees ⁇ 0.2 degree 2-theta.
  • the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at two or more of 9.9, 13.3, 18.5, 19.4, 20.0, 22.6, and 24.7 degrees ⁇ 0.2 degrees 2-theta.
  • the choline salt of Formula I can be characterized by a DSC thermogram substantially as shown in Figure 9. As Figure 9 shows, the choline salt of Formula I produced an endothermic peak at 157.97 °C, with a peak onset temperature of 148.62 °C, and an enthalpy of melting of 22.76 J/g, when heated at a rate of 10°C/min. In some embodiments of the present disclosure, the choline salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 158°C. In other embodiments of the present disclosure, the choline salt of Formula I is characterized by a DSC enthalpy of melting of about 23 J/g.
  • the choline salt of Formula I is characterized by an XRPD pattern comprising peaks at one or more of 9.9, 13.3, 18.5, 19.4, 20.0, 22.6, and 24.7 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 158°C when heated at a rate of 10°C/min.
  • the choline salt of Formula I is characterized by an TGA profile substantially as shown in Figure 10. As shown in Figure 10, the choline salt of Formula I loses about 4.7% by weight upon heating to 250°C at 20 °C per minute.
  • the disclosure is directed to a benzathine salt of a compound of Formula I, having the formula IB:
  • the disclosure is directed to a crystalline form of the benzathine salt of a compound of Formula I.
  • the benzathine salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the benzathine salt of formula I exhibits an XRPD substantially as shown in Figure 12.
  • the XRPD of the benzathine salt of formula I shown in Figure 12 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 4:
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 4. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 4 above.
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 4 above.
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 4 above. In other aspects, the benzathine salt of Formula I is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 4 above.
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at 5.8, and 18.2 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at 5.8, 16.6, and 18.2 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at 5.8, 16.6, 18.2, and 20.7 degrees ⁇ 0.2 degree 2-theta.
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at 5.8, 12.6, 16.6, 18.2, and 22.2 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at 5.8, 12.6, 16.6, 18.2, and 20.7 degrees ⁇ 0.2 degree 2- theta. . In other embodiments, the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at 5.8, 12.6, 16.6, 18.2, 20.7, and 22.2 degrees ⁇ 0.2 degree 2-theta.
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at two or more of 5.8, 12.6, 16.6, 18.2, 20.7, and 22.2 degrees ⁇ 0.2 degrees 2-theta.
  • the benzathine salt of Formula I can be characterized by a DSC thermogram substantially as shown in Figure 13. As Figure 13 shows, the benzathine salt of Formula I produced an endothermic peak at 111.71 °C, with a peak onset temperature of 108.04 °C, and an enthalpy of melting of 42.55 J/g, when heated at a rate of 10°C/min. In some embodiments of the present disclosure, the benzathine salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 112°C. In other embodiments of the present disclosure, the benzathine salt of Formula I is characterized by a DSC enthalpy of melting of about 43 J/g.
  • the benzathine salt of Formula I is characterized by an XRPD pattern comprising peaks at one or more of 5.8, 12.6, 16.6, 18.2, 20.7, and 22.2 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 112°C when heated at a rate of 10°C/min.
  • the benzathine salt of Formula I is characterized by an TGA profile substantially as shown in Figure 14. As shown in Figure 14, the benzathine salt of Formula I loses about 35.2% by weight upon heating to 300°C at 20 °C per minute.
  • the disclosure is directed to an imidazole salt of a compound of Formula I, having formula IC:
  • the disclosure is directed to a crystalline form of the imidazole salt of a compound of Formula I.
  • the imidazole salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the imidazole salt of Formula I exhibits an XRPD substantially as shown in Figure 16.
  • the XRPD of the imidazole salt of formula I shown in Figure 16 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 5:
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 5. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 5 above.
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 5 above.
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 5 above. In other aspects, the imidazole salt of Formula I is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 5 above.
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at 14.1, and 17.0 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at 14.1, 17.0, 17.9, 18.8, and 20.6 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at 14.1, 17.0, 17.9, 18.8, 20.6, 22.0, 22.9, and 23.8, degrees ⁇ 0.2 degree 2-theta.
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at 6.5, 7.0, 14.1, 17.0, 17.9, 18.8, 20.6, 22.0, 22.9, and 23.8 degrees ⁇ 0.2 degree 2-theta. .
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at 14.1, 17.0, 17.9, 18.8, 20.6, 22.0, 22.9, 23.8, 24.4, and 26.5 degrees ⁇ 0.2 degree 2-theta. .
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at 6.5, 7.0, 14.1, 17.0, 17.9, 18.8, 20.6, 22.0, 22.9, 23.8, 24.4, and 26.5 degrees ⁇ 0.2 degree 2-theta.
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at two or more of 6.5, 7.0, 14.1, 17.0, 17.9, 18.8, 20.6, 22.0, 22.9, 23.8, 24.4, and 26.5 degrees ⁇ 0.2 degrees 2-theta.
  • the imidazole salt of Formula I can be characterized by a DSC thermogram substantially as shown in Figure 17. As Figure 17 shows, the imidazole salt of Formula I produced an endothermic peak at 134.56 °C, with a peak onset temperature of 130.50 °C, and an enthalpy of melting of 9.069 J/g, when heated at a rate of 10°C/min. In some embodiments of the present disclosure, the imidazole salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 135°C. In other embodiments of the present disclosure, the imidazole salt of Formula I is characterized by a DSC enthalpy of melting of about 9.1 J/g.
  • the imidazole salt of Formula I is characterized by an XRPD pattern comprising peaks at one or more of 6.5, 7.0, 14.1, 17.0, 17.9, 18.8, 20.6, 22.0, 22.9, 23.8, 24.4, and 26.5 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 135°C when heated at a rate of 10°C/min.
  • the imidazole salt of Formula I is characterized by an TGA profile substantially as shown in Figure 18. As shown in Figure 18, the imidazole salt of Formula I loses about 4.7% by weight upon heating to 200°C at 20 °C per minute.
  • the disclosure is directed to a piperazine salt of a compound of Formula I, having the formula ID:
  • the disclosure is directed to a crystalline form of the piperazine salt of formula I.
  • the piperazine salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the piperazine salt of formula I exhibits an XRPD substantially as shown in Figure 20.
  • the XRPD of the piperazine salt of formula I shown in Figure 20 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 6:
  • the piperazine salt of Formula 1 (Form 1) (Form 1) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 6. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 6 above.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 6 above.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 6 above. In other aspects, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 6 above.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at 7.1, 12.2, and 14.8 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at 7.1, 12.2, 14.8, and 16.0 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at 7.1, 12.2, 14.8, 16.0, and 17.9 degrees ⁇ 0.2 degree 2-theta.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at 7.1, 12.2, 14.8, 16.0, 17.9, and 19.7 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at 7.1, 12.2, 14.8, 16.0, 17.9, 19.7, and 20.5 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at 7.1, 12.2, 14.8, 16.0, 17.9, 19.7, 20.5, and 22.8 degrees ⁇ 0.2 degree 2-theta.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at two or more of 7.1, 12.2, 14.8, 16.0, 17.9, 19.7, 20.5, and 22.8 degrees ⁇ 0.2 degrees 2-theta.
  • the piperazine salt of Formula 1 (Form 1) can be characterized by a DSC thermogram substantially as shown in Figure 21. As Figure 21 shows, the piperazine salt of Formula 1 (Form 1) produced an endothermic peak at 160.50 °C, with a peak onset temperature of 150.65 °C, and an enthalpy of melting of 39.04 J/g, when heated at a rate of 10°C/min.
  • the piperazine salt of Formula 1 (Form 1) is characterized by a DSC thermogram comprising an endothermic peak at about 160°C. In other embodiments of the present disclosure, the piperazine salt of Formula 1 (Form 1) is characterized by a DSC enthalpy of melting of about 39 J/g.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an XRPD pattern comprising peaks at one or more of 7.1, 12.2, 14.8, 16.0, 17.9, 19.7, 20.5, and 22.8 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 160°C when heated at a rate of 10°C/min.
  • the piperazine salt of Formula 1 (Form 1) is characterized by an TGA profile substantially as shown in Figure 22. As shown in Figure 22, the piperazine salt of Formula 1 (Form 1) loses about 14.3% by weight upon heating to 300°C at 20 °C per minute.
  • the piperazine salt of Formula I (Form 2) exhibits an XRPD substantially as shown in Figure 20A.
  • the XRPD of the piperazine salt of Formula I (Form 2) shown in Figure 20A comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 6A:
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 6A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 6A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 6A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 6A above.
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 6 A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 6A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 6A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 6A above.
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 6A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 6 A above. In other aspects, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 6A above.
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 16.5, and 17.8 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 5.5, 6.2, 8.6, 14.0, 16.5, and 17.8, degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 16.5, 17.8, 19.1, 20.5, 22.1, and 23.0 degrees ⁇ 0.2 degree 2-theta.
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 5.5, 6.2, 8.6, 14.0, 16.5, 17.8, 19.1, and 20.5 degrees ⁇ 0.2 degree 2-theta. .
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 8.6, 14.0, 16.5, 17.8, 19.1, 20.5, 22.1, and 23.0 degrees ⁇ 0.2 degree 2-theta. .
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 5.5, 6.2, 8.6, 14.0, 16.5, 17.8, 19.1, 20.5, 22.1, and 23.0 degrees ⁇ 0.2 degree 2-theta.
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at two or more of 5.5, 6.2, 8.6, 14.0, 16.5, 17.8, 19.1, 20.5, 22.1, and 23.0 degrees ⁇ 0.2 degrees 2-theta.
  • the piperazine salt of Formula I (Form 2) can be characterized by a DSC thermogram substantially as shown in Figure 21A.
  • Figure 21 A shows, the piperazine salt of Formula I (Form 2) produced an endothermic peak at 142.60 °C, with a peak onset temperature of 139.29 °C, and an enthalpy of melting of 6.904 J/g, when heated at a rate of 10°C/min.
  • the piperazine salt of Formula I (Form 2) is characterized by a DSC thermogram comprising an endothermic peak at about 143°C.
  • the piperazine salt of Formula I (Form 2) is characterized by a DSC enthalpy of melting of about 6.9 J/g.
  • the piperazine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at one or more of 5.5, 6.2, 8.6, 14.0, 16.5, 17.8, 19.1, 20.5, 22.1, and 23.0 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 143°C when heated at a rate of 10°C/min.
  • the piperazine salt of Formula I (Form 3) exhibits an XRPD substantially as shown in Figure 20B.
  • the XRPD of the piperazine salt of Formula I (Form 3) shown in Figure 20B comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 6B:
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 6B. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 6B above.
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 6B above.
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 6B above. In other aspects, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 6B above.
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at 18.5, 19.4, and 19.9 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at 16.5, 16.9, 18.5, 19.4, 19.9, and 22.7 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at 13.8, 16.5, 16.9, 18.5, 19.4, 19.9, and 22.7 degrees ⁇ 0.2 degree 2-theta.
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at 11.6, 13.8, 16.5, 16.9, 18.5, 19.4, and 19.9 degrees ⁇ 0.2 degree 2- theta. .
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at 11.6, 13.8, 16.5, 16.9, 18.5, 19.4, 19.9, and 22.7 degrees ⁇
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at 6.3, 6.7, 11.0, 11.6, 13.8, 16.5, 16.9, 18.5, 19.4, 19.9, and 22.7 degrees ⁇ 0.2 degree 2-theta.
  • the piperazine salt of Formula I (Form 3) is characterized by an XRPD pattern comprising peaks at two or more of 6.3, 6.7, 11.0, 11.6, 13.8, 16.5, 16.9, 18.5, 19.4, 19.9, and 22.7 degrees ⁇ 0.2 degrees 2-theta.
  • the disclosure is directed to a piperidine salt of a compound of Formula I, having the formula IE:
  • the disclosure is directed to a crystalline form of the piperidine salt of formula I.
  • the piperidine salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the piperidine salt of Formula I (Form 1) exhibits an XRPD substantially as shown in Figure 24.
  • the XRPD of the piperidine salt of Formula I (Form 1) shown in Figure 24 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 7:
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 7. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 7 above.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 7 above.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 7 above. In other aspects, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 7 above.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 7.3, and 17.9 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 16.1, and 17.9 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 14.3, 14.8, 16.1, and 17.9 degrees ⁇ 0.2 degree 2-theta.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 14.3, 14.8, 16.1, 17.9, and 19.8 degrees ⁇ 0.2 degree 2-theta. .
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 14.3, 14.8, 16.1, 17.9, 19.8, and 20.6 degrees ⁇ 0.2 degree 2-theta.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 14.3, 14.8, 16.1, 17.9, 19.8, 20.6, and 22.9 degrees ⁇ 0.2 degree 2-theta.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at two or more of 7.3, 12.2, 14.3, 14.8, 16.1, 17.9, 19.8, 20.6, and 22.9 degrees ⁇ 0.2 degrees 2-theta.
  • the piperidine salt of Formula I (Form 1) can be characterized by a DSC thermogram substantially as shown in Figure 25. As Figure 25 shows, the piperidine salt of Formula I (Form 1) produced an endothermic peak at 174.17 °C, with a peak onset temperature of 161.09 °C, and an enthalpy of melting of 59.20 J/g, when heated at a rate of 10°C/min.
  • the piperidine salt of Formula I (Form 1) is characterized by a DSC thermogram comprising an endothermic peak at about 174°C. In other embodiments of the present disclosure, the piperidine salt of Formula I (Form 1) is characterized by a DSC enthalpy of melting of about 59 J/g.
  • the piperidine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at one or more of 7.3, 12.2, 14.3, 14.8, 16.1, 17.9, 19.8, 20.6, and 22.9 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 174°C when heated at a rate of 10°C/min.
  • the piperidine salt of Formula I (Form 1) is characterized by an TGA profile substantially as shown in Figure 26. As shown in Figure 26, the piperidine salt of Formula I (Form 1) loses about 17.6% by weight upon heating to 300°C at 20 °C per minute.
  • the piperidine salt of Formula I (Form 2) exhibits an XRPD substantially as shown in Figure 24A.
  • the XRPD of the piperidine salt of Formula I (Form 2) shown in Figure 24A comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 7A:
  • the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 7A. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 7A above.
  • the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 7A above.
  • the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 7A above. In other aspects, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 7A above.
  • the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising a peak at 18.3 degrees ⁇ 0.2 degree 2-theta. In other embodiments, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 16.8, and 18.3 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 10.9, 16.8, and 18.3 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 16.8, 18.3, and 20.7 degrees ⁇ 0.2 degree 2-theta.
  • the piperidine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at two or more of 10.9, 16.8, 18.3, and 20.7 degrees ⁇ 0.2 degrees 2-theta.
  • the disclosure is directed to a potassium salt of a compound of Formula I, having the formula IF:
  • the potassium salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the potassium salt of formula I exhibits an XRPD substantially as shown in Figure 28.
  • the XRPD of the potassium salt of formula I shown in Figure 28 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 8: Table 8.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 8. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 8 above.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 8 above.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 8 above. In other aspects, the potassium salt of Formula I is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 8 above.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at 9.1, 10.4, 18.0, and 19.3 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at 10.4, 18.0, 19.3, 22.8, and 24.4 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at 9.1, 10.4, 19.3, and 22.8 degrees ⁇ 0.2 degree 2-theta.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at 9.1, 10.4, 18.0, 19.3, and 24.4 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at 9.1, 10.4, 18.0, 19.3, 22.8, and 24.4 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at 9.1, 10.4, 15.1, 18.0, 19.3, 22.8, and 24.4 degrees ⁇ 0.2 degree 2-theta.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at two or more of 9.1, 10.4, 12.5, 15.1, 18.0, 19.3, 22.8, and 24.4 degrees ⁇ 0.2 degrees 2-theta.
  • the potassium salt of Formula I can be characterized by a DSC thermogram substantially as shown in Figure 29. As Figure 29 shows, the potassium salt of Formula I produced an endothermic peak at 149.53 °C, with a peak onset temperature of 135.10 °C, and an enthalpy of melting of 45.20 J/g, when heated at a rate of 10°C/min.
  • the potassium salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 150°C.
  • the potassium salt of Formula I is characterized by a DSC enthalpy of melting of about 45 J/g.
  • the potassium salt of Formula I is characterized by an XRPD pattern comprising peaks at one or more of9.1, 10.4, 12.5, 15.1, 18.0, 19.3, 22.8, and 24.4 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 150°C when heated at a rate of 10°C/min.
  • the disclosure is directed to a (S)-(-)- ⁇ -Methylbenzylamine salt of a compound of Formula I, having the formula IG: (IG).
  • the disclosure is directed to a crystalline form of the (S)-(-)- ⁇ - methylbenzylamine salt of Formula I.
  • the (S)-(-)- ⁇ -methylbenzylamine salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of formula I exhibits an
  • the XRPD of the (S)-(-)- ⁇ -Methylbenzylamine salt of formula I shown in Figure 30 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2- theta), line spacings (d values), and relative intensities as shown in Table 9:
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 9.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ - Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ - Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ - Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ - Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 9 above.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising a peaks at 18.2 degrees ⁇ 0.2 degrees 2-theta.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising a peak at 19.9 degrees ⁇ 0.2 degrees 2-theta.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising peaks at 18.2 and 19.9 degrees ⁇ 0.2 degree 2-theta.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I can be characterized by a DSC thermogram substantially as shown in Figure 31.
  • Figure 31 shows, the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I produced an endothermic peak at 75.30 °C, with a peak onset temperature of 47.77 °C, and an enthalpy of melting of 106.3 J/g, followed by an endothermic peak at 113.73 °C, with a peak onset temperature of 108.86 °C, and an enthalpy of melting of 16.39 J/g when heated at a rate of 10°C/min.
  • the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 75°C. In other embodiments of the present disclosure, the (S)-(-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by a DSC enthalpy of melting of about 106.3 J/g. In some embodiments of the present disclosure, the (S)- (-)- ⁇ -Methylbenzylamine salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 114°C. In other embodiments of the present disclosure, the (S)-(-)- ⁇ - Methylbenzylamine salt of Formula I is characterized by a DSC enthalpy of melting of about 16.4 J/g.
  • the (S)-(-)- ⁇ - Methylbenzylamine salt of Formula I is characterized by an XRPD pattern comprising peaks at one or more of 18.2 and 19.9 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 75°C or at about 114 °C when heated at a rate of 10°C/min.
  • the disclosure is directed to an ethylenediamine salt of the compound of Formula I, having formula IH:
  • the disclosure is directed to a crystalline form of an ethylenediamine salt of the compound of Formula I.
  • the ethylenediamine salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the ethylenediamine salt of formula I exhibits an
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 10. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 10 above.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 10 above.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 10 above. In other aspects, the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 10 above.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising a peak at 9.4, 10.6, 17.7, and 18.3 degrees ⁇ 0.2 degrees 2-theta.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 9.4, 10.6, 15.4, 17.7, and 18.3 degrees ⁇ 0.2 degrees 2-theta.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 9.4, 10.6, 15.4, 17.7, 18.3, and 19.6 degrees ⁇ 0.2 degree 2- theta.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 9.4, 10.6, 15.4, 17.7, 18.3, 19.6, and 22.0 degrees ⁇ 0.2 degree 2-theta. .
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 9.4, 10.6, 15.4, 17.7, 18.3, 19.6, 22.0, and 23.1 degrees ⁇ 0.2 degree 2-theta. .
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at 9.4, 10.6, 15.4, 17.7, 18.3, 19.6, 22.0, 23.1, and 24.8 degrees ⁇ 0.2 degree 2-theta.
  • the ethylenediamine salt of Formula I (Form 1) is characterized by an XRPD pattern comprising peaks at two or more of 9.4, 10.6,
  • the ethylenediamine salt of formula I exhibits an
  • the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 11. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 11 above.
  • the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 11 above.
  • the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 11 above. In other aspects, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 11 above.
  • the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising a peak at 17.8 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 17.8, and 21.8 degrees ⁇ 0.2 degrees 2-theta. In other embodiments, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 17.8, 21.8, and 22.7 degrees ⁇ 0.2 degree 2-theta.
  • the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 17.8, 21.8, 22.7, and 25.9 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 17.8, 21.8, 22.7, 25.9, and 29.5 degrees ⁇ 0.2 degree 2-theta. . In other embodiments, the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at 17.8, 21.8, 22.7, 25.9, 29.5, and 35.7 degrees ⁇ 0.2 degree 2-theta.
  • the ethylenediamine salt of Formula I (Form 2) is characterized by an XRPD pattern comprising peaks at two or more of 17.8, 21.8, 22.7, 25.9, 29.5, and 35.7 degrees ⁇ 0.2 degrees 2-theta.
  • the disclosure is directed to a 4-((2-aminoethyl)amino)-4- methylpentan-2-one salt of a compound of Formula I, having formula IK:
  • the disclosure is directed to a crystalline form of the 4-((2- aminoethyl)amino)-4-methylpentan-2-one salt of Formula I.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is substantially free of any other salt or solid form of Formula I.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of formula I exhibits an XRPD substantially as shown in Figure 34.
  • the XRPD of the 4-((2- aminoethyl)amino)-4-methylpentan-2-one salt of formula I shown in Figure 34 comprises reflection angles (degrees 2-theta ⁇ 0.2 degrees 2-theta), line spacings (d values), and relative intensities as shown in Table 12:
  • the 4-((2-aminoethyl)amino)-4- methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising a peak at one of the angles listed in Table 12.
  • the 4-((2-aminoethyl)amino)-4- methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising more than one peak at one of the angles listed in Table 12 above.
  • the 4-((2- aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising two peaks selected from the angles listed in Table 12 above.
  • the 4- ((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising three peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising four peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising five peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising six peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising seven peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising eight peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan- 2-one salt of Formula I is characterized by an XRPD pattern comprising nine peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4- methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising ten peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)- 4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising more than ten peaks selected from the angles listed in Table 12 above.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising peaks at 16.3, 17.2, and 18.0 degrees ⁇ 0.2 degrees 2-theta.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2- one salt of Formula I is characterized by an XRPD pattern comprising peaks at 12.2, 12.8, 16.3, 17.2, 18.0, and 20.8 degrees ⁇ 0.2 degrees 2-theta.
  • the 4-((2- aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising peaks at 16.3, 17.2, 18.0, 20.8, 23.2, 24.3, and 26.6 degrees ⁇ 0.2 degree 2-theta.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 12.8, 16.3, and 17.2 degrees ⁇ 0.2 degree 2-theta. .
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 12.8, 16.3, 17.2, 18.0, 20.8, and 23.2 degrees ⁇ 0.2 degree 2-theta.
  • the 4-((2- aminoethyl)amino)-4-methylpentan-2-one salt f Formula I is characterized by an XRPD pattern comprising peaks at 7.3, 12.2, 12.8, 16.3, 17.2, 18.0, 20.8, 23.2, 24.3, and 26.6 degrees ⁇ 0.2 degree 2-theta.
  • the 4-((2-aminoethyl)amino)-4- methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising peaks at two or more of 7.3, 12.2, 12.8, 16.3, 17.2, 18.0, 20.8, 23.2, 24.3, and 26.6 degrees ⁇ 0.2 degrees 2-theta.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I can be characterized by a DSC thermogram substantially as shown in Figure 35.
  • Figure 35 shows, the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I produced an endothermic peak at 170.34 °C, with a peak onset temperature of 161.07 °C, and an enthalpy of melting of 41.18 J/g, when heated at a rate of 10°C/min.
  • the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by a DSC thermogram comprising an endothermic peak at about 170°C. In other embodiments of the present disclosure, the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I is characterized by a DSC enthalpy of melting of about 41 J/g.
  • the 4-((2-aminoethyl)amino)- 4-methylpentan-2-one salt of Formula I is characterized by an XRPD pattern comprising peaks at one or more of 7.3, 12.2, 12.8, 16.3, 17.2, 18.0, 20.8, 23.2, 24.3, and 26.6 degrees ⁇ 0.2 degrees 2-theta, and a DSC thermogram comprising an endothermic peak at about 170°C when heated at a rate of 10°C/min.
  • the 4-((2-aminoethyl)amino)- 4-methylpentan-2-one salt of Formula I is characterized by an TGA profile substantially as shown in Figure 36. As shown in Figure 36, the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I loses about 13.5% by weight upon heating to 250°C at 20 °C per minute.
  • compositions and methods of administration are provided.
  • compositions are typically formulated to provide a therapeutically effective amount of a compound of the present disclosure as the active ingredient, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • the pharmaceutical compositions contain pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • pharmaceutically acceptable excipients including inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • compositions can be administered alone or in combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions.
  • the one or more compounds of the invention and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time.
  • the concentration of one or more compounds provided in the pharmaceutical compositions of the present invention is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% (or a number in the range defined by and including any two numbers above)
  • the concentration of one or more compounds of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25%, 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25%, 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25%, 13%, 12.75%, 12.50%, 12.25%, 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25%, 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25%, 7%, 6.75%, 6.50%, 6.25%, 6%, 5.75%, 5.50%, 5.25%, 5%, 5%,
  • the concentration of one or more compounds of the invention is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40%, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v.
  • the concentration of one or more compounds of the invention is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
  • the amount of one or more compounds of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g,
  • the amount of one or more compounds of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g,
  • the amount of one or more compounds of the invention is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g.
  • the compounds according to the invention are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used. An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • a pharmaceutical composition of the invention typically contains an active ingredient (i.e., a compound of the disclosure) of the present invention or a pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including but not limited to inert solid diluents and fillers, diluents, sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • an active ingredient i.e., a compound of the disclosure
  • a pharmaceutically acceptable salt and/or coordination complex thereof include but not limited to inert solid diluents and fillers, diluents, sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • compositions for oral administration are provided.
  • the invention provides a pharmaceutical composition for oral administration containing a compound of the invention, and a pharmaceutical excipient suitable for oral administration.
  • the invention provides a solid pharmaceutical composition for oral administration containing: (i) an effective amount of a compound of the invention; optionally (ii) an effective amount of a second agent; and (iii) a pharmaceutical excipient suitable for oral administration.
  • the composition further contains: (iv) an effective amount of a third agent.
  • the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption.
  • Pharmaceutical compositions of the invention suitable for oral administration can be presented as discrete dosage forms, such as capsules, cachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in- water emulsion, or a water-in-oil liquid emulsion.
  • Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient into association with the carrier, which constitutes one or more necessary ingredients.
  • compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free- flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising an active ingredient, since water can facilitate the degradation of some compounds.
  • water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf- life or the stability of formulations over time.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms of the invention which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained.
  • anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits.
  • suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
  • An active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier can take a wide variety of forms depending on the form of preparation desired for administration.
  • any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro- crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose.
  • suitable carriers include powders, capsules, and tablets, with the solid oral preparations. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, com starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
  • suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Disintegrants may be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which may disintegrate in the bottle. Too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art.
  • Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
  • Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, com oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof.
  • a lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
  • the active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
  • the tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
  • a suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10.
  • An empirical parameter used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance ("HLB" value).
  • HLB hydrophilic-lipophilic balance
  • Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
  • Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.
  • lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10.
  • HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
  • Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di -glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycer
  • ionic surfactants include, by way of example: lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and diglycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • Ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG- phosphatidylethanolamine, PVP -phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, capry
  • Hydrophilic non-ionic surfactants may include, but are not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; poly glycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene glycol sorb
  • hydrophilic-non-ionic surfactants include, without limitation, PEG- 10 laurate, PEG- 12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG- 12 oleate, PEG- 15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG- 15 stearate, PEG-32 distearate, PEG-40 stearate, PEG- 100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyce
  • Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxy ethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/ vitamin derivatives; and mixtures thereof.
  • preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
  • the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present invention and to minimize precipitation of the compound of the present invention. This can be especially important for compositions for non-oral use, e.g., compositions for injection.
  • a solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
  • solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether (glycofurol) or methoxy PEG ; amides and other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone,
  • solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
  • the amount of solubilizer that can be included is not particularly limited.
  • the amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art.
  • the solubilizer can be in a weight ratio of 10%, 25 %o, 50%), 100%o, or up to about 200%> by weight, based on the combined weight of the drug, and other excipients.
  • solubilizer may also be used, such as 5%>, 2%>, 1%) or even less.
  • the solubilizer may be present in an amount of about 1%> to about 100%, more typically about 5%> to about 25%> by weight.
  • the composition can further include one or more pharmaceutically acceptable additives and excipients.
  • additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
  • an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons.
  • pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like.
  • bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thiogly colic acid, toluenesulfonic acid, uric acid, and the like.
  • a pharmaceutically acceptable acid such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids
  • Salts of polyprotic acids such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used.
  • the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like.
  • Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium.
  • Suitable acids are pharmaceutically acceptable organic or inorganic acids. Examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like.
  • suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p- toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thiogly colic acid, toluenesulfonic acid, uric acid and the like.
  • compositions for injection are provided.
  • the invention provides a pharmaceutical composition for injection containing a compound of the present invention and a pharmaceutical excipient suitable for injection.
  • a pharmaceutical composition for injection containing a compound of the present invention and a pharmaceutical excipient suitable for injection.
  • Components and amounts of agents in the compositions are as described herein.
  • the forms in which the novel compositions of the present invention may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, com oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
  • Aqueous solutions in saline are also conventionally used for injection.
  • Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • Sterile injectable solutions are prepared by incorporating the compound of the present invention in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • certain desirable methods of preparation are vacuum-drying and freeze- drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • compositions for topical e.g. transdermal delivery.
  • the invention provides a pharmaceutical composition for transdermal delivery containing a compound of the present invention and a pharmaceutical excipient suitable for transdermal delivery.
  • Compositions of the present invention can be formulated into preparations in solid, semisolid, or liquid forms suitable for local or topical administration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions.
  • DMSO dimethylsulfoxide
  • carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients.
  • a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
  • compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum comeum permeability barrier of the skin.
  • suitable solid or gel phase carriers or excipients which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum comeum permeability barrier of the skin.
  • penetration- enhancing molecules known to those trained in the art of topical formulation.
  • humectants e.g., urea
  • glycols e.g., propylene glycol
  • alcohols e.g., ethanol
  • fatty acids e.g., oleic acid
  • surfactants e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.glycerol monolaurate, sulfoxides, terpenes (e.g., menthol)
  • amines amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • transdermal delivery devices patches
  • Such transdermal patches may be used to provide continuous or discontinuous infusion of a compound of the present invention in controlled amounts, either with or without another agent.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • compositions for inhalation are provided.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be atached to a face mask tent, or intermitent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.
  • compositions may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art.
  • Administration of the compounds or pharmaceutical composition of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. Compounds can also be administered intraadiposally or intrathecally. [00234] In some embodiments, the compounds or pharmaceutical composition of the present invention are administered by intravenous injection.
  • an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. by dividing such larger doses into several small doses for administration throughout the day.
  • a compound of the invention is administered in a single dose.
  • administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly.
  • injection e.g., intravenous injection
  • other routes may be used as appropriate.
  • a single dose of a compound of the invention may also be used for treatment of an acute condition.
  • a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • Administration of the compounds of the invention may continue as long as necessary.
  • a compound of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days.
  • a compound of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day.
  • a compound of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
  • compositions of the invention may also be delivered via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
  • a method of administration may, for example, aid in the prevention or amelioration of restenosis following procedures such as balloon angioplasty.
  • compounds of the invention may slow or inhibit the migration and proliferation of smooth muscle cells in the arterial wall which contribute to restenosis.
  • a compound of the invention may be administered, for example, by local delivery from the struts of a stent, from a stent graft, from grafts, or from the cover or sheath of a stent.
  • a compound of the invention is admixed with a matrix.
  • Such a matrix may be a polymeric matrix, and may serve to bond the compound to the stent.
  • Polymeric matrices suitable for such use include, for example, lactone-based polyesters or copolyesters such as polylactide, polycaprolactonglycolide, polyorthoesters, polyanhydrides, polyaminoacids, polysaccharides, polyphosphazenes, poly (ether-ester) copolymers (e.g. PEO-PLLA); polydimethylsiloxane, poly(ethylene-vinylacetate), acrylate-based polymers or copolymers (e.g.
  • Compounds of the invention may be applied to the surface of the stent by various methods such as dip/spin coating, spray coating, dip-coating, and/or brush-coating.
  • the compounds may be applied in a solvent and the solvent may be allowed to evaporate, thus forming a layer of compound onto the stent.
  • the compound may be located in the body of the stent or graft, for example in microchannels or micropores.
  • stents When implanted, the compound diffuses out of the body of the stent to contact the arterial wall.
  • stents may be prepared by dipping a stent manufactured to contain such micropores or microchannels into a solution of the compound of the invention in a suitable solvent, followed by evaporation of the solvent. Excess drug on the surface of the stent may be removed via an additional brief solvent wash.
  • compounds of the invention may be covalently linked to a stent or graft.
  • a covalent linker may be used which degrades in vivo, leading to the release of the compound of the invention. Any bio-labile linkage may be used for such a purpose, such as ester, amide or anhydride linkages.
  • Compounds of the invention may additionally be administered intravascularly from a balloon used during angioplasty. Extravascular administration of the compounds via the peri card or via advential application of formulations of the invention may also be performed to decrease restenosis.
  • the compounds of the invention may be administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure. [00244] When a compound of the invention is administered in a composition that comprises one or more agents, and the agent has a shorter half- life than the compound of the invention unit dose forms of the agent and the compound of the invention may be adjusted accordingly.
  • the subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • the method typically comprises administering to a subject a therapeutically effective amount of a compound of the invention.
  • the therapeutically effective amount of the subject combination of compounds may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • IC 50 refers to the half maximal inhibitory concentration of an inhibitor in inhibiting biological or biochemical function. This quantitative measure indicates how much of a particular inhibitor is needed to inhibit a given biological process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half. In other words, it is the half maximal (50%) inhibitory concentration (IC) of a substance (50% IC, or IC50). EC50 refers to the plasma concentration required for obtaining 50%> of a maximum effect in vivo. [00249] In some embodiments, the subject methods utilize a MCL-1 inhibitor with an IC50 value of about or less than a predetermined value, as ascertained in an in vitro assay.
  • the MCL-1 inhibitor inhibits MCL-1 a with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less,
  • nM or less 190 nM or less, 200 nM or less, 225 nM or less, 250 nM or less, 275 nM or less, 300 nM or less,
  • nM or less 475 nM or less, 500 nM or less, 550 nM or less, 600 nM or less, 650 nM or less, 700 nM or less,
  • the MCL-1 inhibitor selectively inhibits MCL-1 a with an IC50 value that is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, or 1000 times less (or a number in the range defined by and including any two numbers above)than its IC50 value against one, two, or three other MCL-ls.
  • the MCL-1 inhibitor selectively inhibits MCL-1 a with an IC50 value that is less than about 1 nM, 2 nM, 5 nM, 7 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 120 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 225 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900
  • MCL-1 Any disease condition that results directly or indirectly from an abnormal activity or expression level of MCL-1 can be an intended disease condition.
  • MCL-1 has been implicated, for example, auto-immune diseases, neurodegeneration (such as Parkinson’s disease, Alzheimer’s disease and ischaemia), inflammatory diseases, viral infections and cancer such as, for example, colon cancer, breast cancer, small-cell lung cancer, non-small-cell lung cancer, bladder cancer, ovarian cancer, prostate cancer, chronic lymphoid leukemia, lymphoma, myeloma, acute myeloid leukemia, or pancreatic cancer.
  • auto-immune diseases such as Parkinson’s disease, Alzheimer’s disease and ischaemia
  • inflammatory diseases such as, for example, colon cancer, breast cancer, small-cell lung cancer, non-small-cell lung cancer, bladder cancer, ovarian cancer, prostate cancer, chronic lymphoid leukemia, lymphoma, myeloma, acute myeloid leukemia, or pancreatic cancer.
  • Non- limiting examples of such conditions include but are not limited to Acanthoma, Acinic cell carcinoma, Acoustic neuroma, Acral lentiginous melanoma, Acrospiroma, Acute eosinophilic leukemia, Acute lymphoblastic leukemia, Acute lymphocytic leukemia, Acute megakaryoblastic leukemia, Acute monocytic leukemia, Acute myeloblasts leukemia with maturation, Acute myeloid dendritic cell leukemia, Acute myeloid leukemia, Acute myelogenous leukemia, Acute promyelocytic leukemia, Adamantinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenoma, Adenomatoid odontogenic tumor, Adrenocortical carcinoma, Adult T-cell leukemia, Aggressive NK-cell leukemia, AIDS-Related Cancers, AIDS-related lymphoma, Alveolar
  • said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy, retinopathy of prematurity
  • age-related macular degeneration hemangio
  • said method is for treating a disease selected from breast cancer, lung cancer, pancreatic cancer, prostate cancer, colon cancer, ovarian cancer, uterine cancer, or cervical cancer.
  • said method is for treating a disease selected from leukemia such as acute myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, hairy cell leukemia, myelodysplasia, myeloproliferative disorders, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), mastocytosis, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), myelodysplastic syndrome (MDS) or epidermoid cancer.
  • AML acute myeloid leukemia
  • AML acute lymphocytic leukemia
  • chronic lymphocytic leukemia chronic myeloid leukemia
  • CML chronic myelogenous leukemia
  • mastocytosis chronic lymphocytic leukemia
  • CLL multiple myeloma
  • MDS myelodysplastic syndrome
  • Compounds of the disclosure, as well as pharmaceutical compositions comprising them, can be administered to treat any of the described diseases, alone or in combination with a medical therapy.
  • Medical therapies include, for example, surgery and radiotherapy (e.g gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, systemic radioactive isotopes).
  • compounds of the disclosure as well as pharmaceutical compositions comprising them, can be administered to treat any of the described diseases, alone or in combination with one or more other agents.
  • the compounds of the disclosure as well as pharmaceutical compositions comprising them, can be administered in combination with agonists of nuclear receptors agents.
  • the compounds of the disclosure as well as pharmaceutical compositions comprising them, can be administered in combination with antagonists of nuclear receptors agents.
  • the compounds of the disclosure as well as pharmaceutical compositions comprising them, can be administered in combination with an anti-proliferative agent.
  • the compounds of the invention can be used in combination with chemotherapeutic agents, agonists or antagonists of nuclear receptors, or other anti-proliferative agents.
  • the compounds of the invention can also be used in combination with a medical therapy such as surgery or radiotherapy, e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes.
  • chemotherapeutic agents include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, all-trans retinoic acid, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bendamustine, bevacizumab, bexarotene, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin difti
  • the compounds of the invention can be used in combination with a therapeutic agent that targets an epigenetic regulator.
  • epigenetic regulators include bromodomain inhibitors, the histone lysine methyltransferase inhibitors, histone arginine methyl transferase inhibitors, histone demethylase inhibitors, histone deacetylase inhibitors, histone acetylase inhibitors, and DNA methyltransferase inhibitors.
  • Histone deacetylase inhibitors include, e.g., vorinostat.
  • Histone arginine methyl transferase inhibitors include inhibitors of protein arginine methyltransferases (PRMTs) such as PRMT5, PRMTl and PRMT4.
  • DNA methyltransferase inhibitors include inhibitors of DNMT1 and DNMT3.
  • the compounds of the invention can be used in combination with targeted therapies, including JAK kinase inhibitors (e.g. Ruxolitinib), PI3 kinase inhibitors including PI3K-delta selective and broad spectrum PI3K inhibitors, MEK inhibitors, Cyclin Dependent kinase inhibitors, including CDK4/6 inhibitors and CDK9 inhibitors, BRAF inhibitors, mTOR inhibitors, proteasome inhibitors (e.g. Bortezomib, Carfilzomib), HD AC inhibitors (e.g.
  • JAK kinase inhibitors e.g. Ruxolitinib
  • PI3 kinase inhibitors including PI3K-delta selective and broad spectrum PI3K inhibitors
  • MEK inhibitors Cyclin Dependent kinase inhibitors
  • CDK4/6 inhibitors and CDK9 inhibitors including CDK4/6 inhibitors and CDK9 inhibitors
  • BRAF inhibitors e.g. Bortez
  • panobinostat panobinostat, vorinostat
  • DNA methyl transferase inhibitors dexamethasone, bromo and extra terminal family member (BET) inhibitors, BTK inhibitors (e.g. ibrutinib, acalabrutinib), BCL2 inhibitors (e.g. venetoclax), dual BCL2 family inhibitors (e.g. BCL2/BCLxL), PARP inhibitors, FLT3 inhibitors, or LSD1 inhibitors.
  • BET bromo and extra terminal family member
  • BTK inhibitors e.g. ibrutinib, acalabrutinib
  • BCL2 inhibitors e.g. venetoclax
  • dual BCL2 family inhibitors e.g. BCL2/BCLxL
  • PARP inhibitors FLT3 inhibitors, or LSD1 inhibitors.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody.
  • the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475), or PDR001.
  • the anti-PD-1 monoclonal antibody is nivolumab or pembrolizumab.
  • the anti-PDl antibody is pembrolizumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-Ll monoclonal antibody.
  • the anti-PD-Ll monoclonal antibody is atezolizumab, durvalumab, or BMS-935559.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab.
  • the agent is an alkylating agent, a proteasome inhibitor, a corticosteroid, or an immunomodulatory agent.
  • an alkylating agent include cyclophosphamide (CY), melphalan (MEL), and bendamustine.
  • the proteasome inhibitor is carfilzomib.
  • the corticosteroid is dexamethasone (DEX).
  • the immunomodulatory agent is lenalidomide (LEN) or pomalidomide (POM).
  • the compound of the invention can be administered in combination with a corticosteroid such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
  • a corticosteroid such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
  • the compound of the invention can be administered in combination with an immune suppressant such as fluocinolone acetonide (Retisert®), rimexolone (AL-2178, Vexol, Alcon), or cyclosporine (Restasis®).
  • an immune suppressant such as fluocinolone acetonide (Retisert®), rimexolone (AL-2178, Vexol, Alcon), or cyclosporine (Restasis®).
  • Step 1 6'-chlorospiro[oxirane-2, l'-tetralin]
  • Step 7 4-[[6-chloro-1-(dimethoxymethyl)tetralin-1-yl]methoxy]-3-nitro-benzenesulfonamide
  • the reaction mixture was quenched with sat. NFUCl (10 mL).
  • the reaction mixture was diluted with water (80 mL) and sat. NFLCl (10 mL), and extracted with EtOAc (100 mL).
  • the organic layer was washed with water (70 mL) and sat. NFLCl (10 mL), and brine (50 mL).
  • the aqueous layers were combined, and back-extracted with EtOAc (60 mL), washed with water (60 mL), and brine (30 mL).
  • Step 8 4-[( 6-chloro-1-formyl-tetralin-1-yl)methoxy]-3-nitro-benzenesulfonamide
  • Step 9 6'-chlorospiro[4, 5-dihydro-2H-1, 5-benzoxazepine-3,1 '-tetralin ]-7-sulfonamide
  • Step 1 4-fluoro-N,N-bis[(4-methoxyphenyl)methyl ]-3-nitro-benzenesulfonamide
  • the mixture was neutralized with 1 N HC1 to pH about 4-5 and diluted with EtOAc (100 mL). The organic layer was separated, washed with 1 N HC1 (10 mL), 7.5% NaHC03 aqueous solution (20 mL), and brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was treated with DCM (30 mL), and hexane was added to the suspension until it became cloudy. The resulting suspension was sonicated for 2 min. and left at r.t. for 1 h. The mixture was filtered, and washed with hexane to afford the desired title product (6.85 g) without further purification. The mother liquid was concentrated under reduced pressure.
  • Step 2 [(lS)-6-chloro-1-(hydroxymethyl)tetralin-1-yljmethyl benzoate and [(1R)-6-chloro-1- (hydroxymethyl)tetralin-1-yl ]methyl benzoate
  • Method A This compound was prepared using procedures analogous to those described for Intermediate 1 using [(lR)-6-chloro-1-formyl-tetralin-1-yl]methyl benzoate to replace the racemic (6-chloro-1-formyl-tetralin-1-yl)methyl benzoate in Step 6.
  • UV length 220 nm, 254 nm, and 280 nm; Column temperature: 30 °C; Back pressure: 120 bar. to afford
  • Step 6 N,N-bis [(4-methoxyphenyl) methyl]-3-nitro-4-[[(1R)-6-chloro-1-formyl-tetralin-1- yl ]methoxy ]benzenesulfonamide
  • Step 7 (S)-6'-chloro-N,N-bis(4-methoxybenzyl)-3',4'-dihydro-2H,2'H- spiro[benzo[b ][1, 4 ]oxazepine-3, 1'-naphthalene ] -7 -sulfonamide
  • Step 8 (3S)-6'-chloro-N,N-bis[(4-methoxyphenyl)methyl]spiro[4,5-dihydro-2H-l,5- benzoxazepine-3, 1 '-tetralin ] -7 -sulfonamide
  • (S)-6'-chloro-N,N-bis(4-methoxybenzyl)-3',4'-dihydro-2H,2'H- spiro[benzo[b][1,4]oxazepine-3,1'-naphthalene]-7-sulfonamide (6.11 g, 6.73 mmol) (crude product from Step 7, 70% purity) in DCM (80 mL) was portion-wise added NaBH(OAc)3 (7.14 g, 33.67 mmol).
  • Step 1 [(1R, 2R)-2-[[(3S)-7-[bis [(4-methoxyphenyl)methyl]sulfamoyl]-6'-chloro-spiro[2,4- dihydro-1,5-benzoxazepine-3, 1 '-tetralin ]-5-yl ]methyl ] cyclobutyl ]methyl acetate
  • Step 2 (3S)-6'-chloro-N ,N-bis[( 4-methoxyphenyl)methyl ]-5-[[(1R, 2R)-2- (hydroxymethyl)cyclobutyl ]methyl ] spiro [2, 4-dihydro-l, 5-benzoxazepine-3, 1 '-tetralin]-7- sulfonamide
  • the solution was stirred at -78 °C for additional 10 min, and allowed to warme slowly to 0 °C. After the starting material was consumed, water (150 mL) was added. The organic layer were separated. The aqueous layer was extracted with DCM (300 mL x 3). The combined organic layers were dried over sodium sulfate and concentrated.
  • Step 4 (3S)-6'-chloro-N ,N-bis[( 4-methoxyphenyl)methyl ]-5-[[(1R, 2R)-2-[( I S)-1- hydroxyallyl ]cyclobutyl ]methyl ]spiro[2, 4-dihydro-l , 5-benzoxazepine-3, 1 '-tetralin ]-7- sulfonamide and ( 3S)-6'-chloro-N,N-bis[( 4-methoxyphenyl)methyl ]-5-[[(1R, 2R)-2-[( 1R)-1- hydroxyallyl ]cyclobutyl ]methyl ]spiro[2, 4-dihydro-l , 5-benzoxazepine-3, 1 '-tetralin ]-7- sulfonamide [00293] Vinylmagnesium bromide (1.0 M solution in THF, 300 mL, 300 mmol) was diluted with THF (
  • Step 5 (3S)-6'-chloro-5-[[(1R, 2R)-2-[( l S)-1-hydroxyallyl ] cyclobutyl ]methyl ] spiro [2, 4-dihydro- 1, 5-benzoxazepine-3, 1 '-tetralin ] -7 -sulfonamide (Intermediate 3)
  • This compound can be prepared by treating ethyl 2-hydroxy -2 -methyl-propanoate with NaH in THF, followed by reaction with allyl bromide. The resulting product is then reacted with sodium hydroxide to give 2-allyloxy -2-methyl-propanoic acid.
  • Step 1 [(lS)-1-[(1R,2R)-2-[[(3S)-7-[(2-allyloxy-2-methyl-propanoyl)sulfamoyl]-6'-chloro- spiro [2, 4-dihydro- 1, 5-benzoxazepine-3, 1 '-tetralin ]-5-yl ]methyl ]cyclobutyl Jallyl ] 2-allyloxy-2- methyl-propanoate
  • Step 2 2-allyloxy-2-methyl-N-[( 3S)-6'-chloro-5-[[( IR 2R)-2-[(lS)-1- hydroxyallyl ]cyclobutyl ]methyl ]spiro[2, 4-dihydro-l , 5-benzoxazepine-3, 1 '-tetralin ]-7- yl ]sulfonyl-propanamide
  • Step 3 (3R, 6R, 7S, 8E, 22S)-6'-chloro-7-hydroxy-l 2, 12-dimethyl-15, 15-dioxo-spiro[ll, 20-dioxa- 15-thia-l, 14-diazatetracyclo[14.7.2.03, 6.019, 24 ]pentacosa-8, 16,18, 24-tetraene-22, 1 '-tetralin ]- 13-one
  • P2 was assigned to (3R,6R,7S,8E,22S)-6'-chloro-7-hydroxy-12,12-dimethyl-15,15- dioxo-spiro[ll,20-dioxa-15-thia-1,14-diazatetracyclo[14.7.2.03,6.019,24]pentacosa-8,16,18,24- tetraene-22,1'-tetralin]-13-one (Example 32).
  • N,N-dimethylcarbamoyl chloride (4.6 mg, 0.04 mmol) was added, and followed by DMAP (5.3 mg, 0.04 mmol).
  • the mixture was stirred at r.t. for 6 h., and diluted with DCM and acidified with 0.5 N HC1 to pH 5-6.
  • the organic phase was separated, washed with water and brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • DSC Figure 2.
  • TGA Figure 3.
  • DVS Figure 4A and 4B.
  • Formula I (23.7 mg (0.036 mmol) amorphous) was added to a 4 mL vial. Methanol (0.4 mL) and water (0.1 mL) were added to give a thin slurry. The mixture was stirred at 50 °C for 3 h to form a slurry. The mixture was cooled to room temperature and stirred for 20 min.
  • Formula I - Form II - Method 1 [00311] Formula I (400 mg; amorphous) was added to a 20 mL vial. Ethanol (7.0 mL) was added to give a slurry. The mixture was stirred at 70 °C for 20 minutes to give a solution. The solution was slowly cooled to give a slurry. The slurry was held over the weekend and then filtered to give Formula I- Form II.
  • Formula I imidazole salt (Formula IC) [00325] Formula I (168.0 mg (0.25 mmol, 1.0 eq.) amorphous) was added to a 25 mL vial. Ethyl acetate (4.0 mL) was added to give a clear solution. 18.9 mg of imidazole (0.275 mmol, 1.1 eq.) was added. The mixture was stirred for 5 minutes to give a clear solution. The mixture was continuously stirred overnight to give a slurry. The mixture was filtered and the cake was dried at room temperature under vacuum overnight to yield 118.0 mg (63.8%) of the imidazole salt of Formula I.
  • DSC Figure 17.
  • DSC Figure 21.
  • TGA Figure 22.
  • DSC Figure 21A.
  • Formula I uiuerazine salt - (Form 3)
  • Formula I (25.0 mg; 0.037 mmol) was added to a 4 mL vial. 0.5 mL of methanol was added and the mixture was stirred for 30 minutes. Piperazine (0.056 mmol, 1.50 eq.) was added and the mixture was stirred for 2 hrs, and then at 50°C for 2 hrs. The mixture was cooled and then stirred at room temperature overnight, and then filtered to give the Formula I piperazine salt.
  • Formula I (25.0 mg; 0.037 mmol) was added to a 4 mL vial. 0.5 mL of THF/methanol was added and the mixture was stirred for 30 minutes. Piperazine (0.056 mmol, 1.50 eq.) was added and the mixture was stirred for 2 hrs, and then at 50°C for 2 hrs. The mixture was cooled and then stirred at room temperature overnight, and then filtered to give the Formula I piperazine salt.
  • DSC Figure 25.
  • TGA Figure 26.
  • the Formula I piperidine salt also was prepared by the reaction of Formula I free acid with 2.0 eq. of piperidine in IPA/MeOH.
  • Formula I 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt
  • Formula I (168.0 mg (0.25 mmol, 1.0 eq.) amorphous) was added to a 25 mL vial. Ethyl acetate (4.0 mL) was added to give a clear solution. 275 pi of 1.0 M ethylene diamine in acetone (0.275 mmol, 1.1 eq.) was added. The mixture was stirred for 5 minutes to give a clear solution. The mixture was continuously stirred overnight to give a slurry. The mixture was filtered and the cake was dried at room temperature under vacuum overnight to yield 102.2 mg (63.8%) of the 4-((2-aminoethyl)amino)-4-methylpentan-2-one salt of Formula I.
  • DSC Figure 35.
  • TGA Figure 36.
  • the Formula I potassium salt was prepared by the reaction of Formula I free acid with potassium hydroxide (2 M in water, 2.0 eq.) in ethanol.
  • DSC Figure 29.
  • the Formula I potassium salt also was prepared by the reaction of Formula I free acid with potassium hydroxide (2 M in water, 2.0 eq.) in isopropanol.
  • Formula I - (S)-(-)- ⁇ methylbenzylamine salt [00364]
  • the Formula I-(S)-(-)- ⁇ -methylbenzylamine salt was prepared by the reaction of Formula I free acid with (S)-(-)- ⁇ -methylbenzylamine (1.5 eq.) in THF/methanol.
  • DSC Figure 31.
  • XRPD patterns can be collected with a PANalytical X'Pert PRO MPD diffractometer using an incident beam of Cu radiation produced using an Optix long, fine-focus source.
  • An elliptically graded multilayer mirror is used to focus Cu K ⁇ X-rays through the specimen and onto the detector.
  • a silicon specimen NIST SRM 640e
  • a specimen of the sample is sandwiched between 3-pm-thick films and analyzed in transmission geometry.
  • a beam-stop, short antiscatter extension, and antiscatter knife edge is used to minimize the background generated by air.
  • Sober slits for the incident and diffracted beams are used to minimize broadening from axial divergence. Diffraction patterns are collected using a scanning position-sensitive detector (X'Celerator) located 240 mm from the specimen and Data Collector software v. 2.2b.
  • X'Celerator scanning position-sensitive detector
  • XRPD patterns also can be collected with a Rigaku MiniFlex X-ray Powder Diffractometer (XRPD) instrument.
  • X-ray radiation is from Copper (Cu) at 1.54056 ⁇ with K b filter.
  • X-ray power 30 KV, 15 mA.
  • TGA Thermogravimetric Analysis
  • DSC Differential Scanning Calorimetry
  • Thermal analysis can be performed using a Mettler Toledo TGA/DSC3+ analyzer. Temperature calibration is performed using phenyl salicylate, indium, tin, and zinc. The sample is placed in an aluminum pan. The sample is sealed, the lid pierced, then inserted into the TG furnace. The furnace is heated under nitrogen.
  • DSC can also be obtained using a TA Instrument Differential Scanning Calorimetry, Model Q20 with autosampler, using a scan rate of 10 °C/min, and nitrogen gas flow at 50 mL/min.
  • TGA can be collected using a TGA Q500 by TA Instruments using a scan rate of 20 °C per minute.
  • the dynamic vapor sorption experiments can be done with a VTI SGA-CxlOO Symmetric Vapor Sorption Analyzer.
  • the moisture uptake profile is completed in three cycles of 10% RH increments with adsorption from 5% to 95% RH, followed by desorption of 10% increments from 95% to 5%.
  • the equilibration criteria are 0.0050 wt% in 5 minutes with a maximum equilibration time of 180 minutes. All adsorption and desorption are performed at room temperature (23-25 °C). No pre-drying step is applied for the samples.
  • the binding affinity of each compound was measured via a fluorescence polarization competition assay, in which the compound competes for the same binding site with the ligand, and thus leads to a dose-dependent anisotropy reduction.
  • the tracer ligand utilized was a fluorescein isothiocyanate labelled peptide (FITC-ARIAQELRRIGDEFNETYTR) derived from Bim (GenScript).
  • the assay was carried out in black half-area 96-well NBS plate (Coming), containing 15 nM of MCL-1 (BPS Bioscience), 5 nM of FITC-Bim and 3-fold serial diluted test compounds in a total volume of 50 ⁇ L of assay buffer (20 mM HEPES, 50 mM NaCl, 0.002% Tween 20, 1 mM TCEP, and 1% DMSO).
  • assay buffer (20 mM HEPES, 50 mM NaCl, 0.002% Tween 20, 1 mM TCEP, and 1% DMSO).
  • the reaction plate was incubated for 1 hour at room temperature.
  • the change of anisotropy is measured with an Envision multimode plate reader (PerkinElmer) at emission wavelength 535 nm.
  • % inhibition 100x(mPDMSO-mP)/(mPDMSO-mPpc), in which mPDMSO is the DMSO control, and mPpc is the positive control.
  • IC 50 values were determined from a 10-point dose response curve by fitting the percent inhibition against compound concentration using the GraphPad Prism software. The inhibition constant K, was subsequently calculated according to the Nikolovska-Coleska’s equation (Anal.
  • Table A Cell free Mcl-l:Bim affinity assay (Mcl-1 Bim) and Cell viability assay (H929_10FBS)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP21730723.0A 2020-05-13 2021-05-13 Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein Pending EP4149947A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063024110P 2020-05-13 2020-05-13
PCT/US2021/032263 WO2021231737A1 (en) 2020-05-13 2021-05-13 Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein

Publications (1)

Publication Number Publication Date
EP4149947A1 true EP4149947A1 (en) 2023-03-22

Family

ID=76306004

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21730723.0A Pending EP4149947A1 (en) 2020-05-13 2021-05-13 Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein

Country Status (11)

Country Link
US (1) US20230357275A1 (ja)
EP (1) EP4149947A1 (ja)
JP (1) JP2023526235A (ja)
KR (1) KR20230024287A (ja)
CN (1) CN115698024A (ja)
AU (1) AU2021271694A1 (ja)
BR (1) BR112022022995A2 (ja)
CA (1) CA3183270A1 (ja)
IL (1) IL298157A (ja)
MX (1) MX2022014219A (ja)
WO (1) WO2021231737A1 (ja)

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3657744A (en) 1970-05-08 1972-04-25 Univ Minnesota Method for fixing prosthetic implants in a living body
US4733665C2 (en) 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5040548A (en) 1989-06-01 1991-08-20 Yock Paul G Angioplasty mehtod
US5061273A (en) 1989-06-01 1991-10-29 Yock Paul G Angioplasty apparatus facilitating rapid exchanges
US5350395A (en) 1986-04-15 1994-09-27 Yock Paul G Angioplasty apparatus facilitating rapid exchanges
US4748982A (en) 1987-01-06 1988-06-07 Advanced Cardiovascular Systems, Inc. Reinforced balloon dilatation catheter with slitted exchange sleeve and method
US4992445A (en) 1987-06-12 1991-02-12 American Cyanamid Co. Transdermal delivery of pharmaceuticals
US5001139A (en) 1987-06-12 1991-03-19 American Cyanamid Company Enchancers for the transdermal flux of nivadipine
CA1322628C (en) 1988-10-04 1993-10-05 Richard A. Schatz Expandable intraluminal graft
US5292331A (en) 1989-08-24 1994-03-08 Applied Vascular Engineering, Inc. Endovascular support device
US5674278A (en) 1989-08-24 1997-10-07 Arterial Vascular Engineering, Inc. Endovascular support device
US6344053B1 (en) 1993-12-22 2002-02-05 Medtronic Ave, Inc. Endovascular support device and method
JP6453507B2 (ja) * 2017-03-30 2019-01-16 アムジエン・インコーポレーテツド Mcl−1タンパク質を阻害する化合物
KR20210089662A (ko) * 2018-11-09 2021-07-16 프렐루드 테라퓨틱스, 인코포레이티드 골수 세포 백혈병-1 (mcl-1) 단백질의 억제제로서 스피로-술폰아미드 유도체

Also Published As

Publication number Publication date
US20230357275A1 (en) 2023-11-09
IL298157A (en) 2023-01-01
CN115698024A (zh) 2023-02-03
KR20230024287A (ko) 2023-02-20
CA3183270A1 (en) 2021-11-18
MX2022014219A (es) 2023-01-16
BR112022022995A2 (pt) 2023-01-17
AU2021271694A1 (en) 2023-01-19
WO2021231737A1 (en) 2021-11-18
JP2023526235A (ja) 2023-06-21

Similar Documents

Publication Publication Date Title
EP3665179B1 (en) Selective inhibitors of protein arginine methyltransferase 5 (prmt5)
US11130769B2 (en) Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (MCL-1) protein
US20220324830A1 (en) CDK Inhibitors And Their Use As Pharmaceuticals
US20220160713A1 (en) Selective inhibitor of protein arginine methyltransferase 5 (prmt5)
US20230212191A1 (en) Spiro-sulfonimidamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein
US20230029094A1 (en) Selective Inhibitors Of Protein Arginine Methyltransferase 5 (PRMT5)
US20230265083A1 (en) Heterocycle CDK Inhibitors And Their Use Thereof
WO2020123994A1 (en) 2-((5-(phenyl)-pyrrolo[2,1-f][1,2,4]triazin-4-yl)amino)-3-(phenyl)propanoic acid derivatives and related compounds as mcl-1 enzyme inhibitors for treating cancer
WO2023056441A1 (en) Cdk inhibitors and their use as pharmaceuticals
US20230357275A1 (en) Spiro-sulfonamide derivatives as inhibitors of myeloid cell leukemia-1 (mcl-1) protein
WO2023178547A1 (en) Polymorphic compounds and uses thereof
US11939340B2 (en) CDK inhibitors and their use as pharmaceuticals
US20240352029A1 (en) CDK Inhibitors And Their Use As Pharmaceuticals
WO2024216249A1 (en) Cdk inhibitors and their use as pharmaceuticals

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221201

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230526

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40090513

Country of ref document: HK